Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Physiol Rev ; 103(2): 1247-1421, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36603156

ABSTRACT

This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.


Subject(s)
Endothelial Cells , Mechanotransduction, Cellular , Humans , Mechanotransduction, Cellular/physiology , Endothelial Cells/metabolism , Epigenesis, Genetic , Signal Transduction/physiology , Myocytes, Smooth Muscle , Stress, Mechanical
2.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34810252

ABSTRACT

Vascular endothelial cells are exposed to shear stresses with disturbed vs. laminar flow patterns, which lead to proinflammatory vs. antiinflammatory phenotypes, respectively. Effective treatment against endothelial inflammation and the consequent atherogenesis requires the identification of new therapeutic molecules and the development of drugs targeting these molecules. Using Connectivity Map, we have identified vitexin, a natural flavonoid, as a compound that evokes the gene-expression changes caused by pulsatile shear, which mimics laminar flow with a clear direction, vs. oscillatory shear (OS), which mimics disturbed flow without a clear direction. Treatment with vitexin suppressed the endothelial inflammation induced by OS or tumor necrosis factor-α. Administration of vitexin to mice subjected to carotid partial ligation blocked the disturbed flow-induced endothelial inflammation and neointimal formation. In hyperlipidemic mice, treatment with vitexin ameliorated atherosclerosis. Using SuperPred, we predicted that apurinic/apyrimidinic endonuclease1 (APEX1) may directly interact with vitexin, and we experimentally verified their physical interactions. OS induced APEX1 nuclear translocation, which was inhibited by vitexin. OS promoted the binding of acetyltransferase p300 to APEX1, leading to its acetylation and nuclear translocation. Functionally, knocking down APEX1 with siRNA reversed the OS-induced proinflammatory phenotype, suggesting that APEX1 promotes inflammation by orchestrating the NF-κB pathway. Animal experiments with the partial ligation model indicated that overexpression of APEX1 negated the action of vitexin against endothelial inflammation, and that endothelial-specific deletion of APEX1 ameliorated atherogenesis. We thus propose targeting APEX1 with vitexin as a potential therapeutic strategy to alleviate atherosclerosis.


Subject(s)
Apigenin/genetics , Apigenin/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Endothelial Cells/metabolism , Active Transport, Cell Nucleus , Animals , Atherosclerosis , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Mice , Phenotype , Phosphorylation , Protein Binding , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , p300-CBP Transcription Factors/metabolism
3.
Proc Natl Acad Sci U S A ; 116(26): 12974-12979, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31182601

ABSTRACT

Pulsatile shear (PS) and oscillatory shear (OS) elicit distinct mechanotransduction signals that maintain endothelial homeostasis or induce endothelial dysfunction, respectively. A subset of microRNAs (miRs) in vascular endothelial cells (ECs) are differentially regulated by PS and OS, but the regulation of the miR processing and its implications in EC biology by shear stress are poorly understood. From a systematic in silico analysis for RNA binding proteins that regulate miR processing, we found that nucleolin (NCL) is a major regulator of miR processing in response to OS and essential for the maturation of miR-93 and miR-484 that target mRNAs encoding Krüppel-like factor 2 (KLF2) and endothelial nitric oxide synthase (eNOS). Additionally, anti-miR-93 and anti-miR-484 restore KLF2 and eNOS expression and NO bioavailability in ECs under OS. Analysis of posttranslational modifications of NCL identified that serine 328 (S328) phosphorylation by AMP-activated protein kinase (AMPK) was a major PS-activated event. AMPK phosphorylation of NCL sequesters it in the nucleus, thereby inhibiting miR-93 and miR-484 processing and their subsequent targeting of KLF2 and eNOS mRNA. Elevated levels of miR-93 and miR-484 were found in sera collected from individuals afflicted with coronary artery disease in two cohorts. These findings provide translational relevance of the AMPK-NCL-miR-93/miR-484 axis in miRNA processing in EC health and coronary artery disease.


Subject(s)
Coronary Artery Disease/genetics , Mechanotransduction, Cellular/genetics , MicroRNAs/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Aged , Animals , Case-Control Studies , Cells, Cultured , Computational Biology , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Gene Knockdown Techniques , Humans , Kruppel-Like Transcription Factors/genetics , Male , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/blood , Middle Aged , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Protein Processing, Post-Translational , RNA Processing, Post-Transcriptional , Serine/metabolism , Stress, Mechanical , Nucleolin
4.
Proc Natl Acad Sci U S A ; 115(1): 133-138, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29255056

ABSTRACT

Leukocyte transmigration across vessel walls is a critical step in the innate immune response. Upon their activation and firm adhesion to vascular endothelial cells (VECs), leukocytes preferentially extravasate across junctional gaps in the endothelial monolayer (paracellular diapedesis). It has been hypothesized that VECs facilitate paracellular diapedesis by opening their cell-cell junctions in response to the presence of an adhering leukocyte. However, it is unclear how leukocytes interact mechanically with VECs to open the VEC junctions and migrate across the endothelium. In this study, we measured the spatial and temporal evolution of the 3D traction stresses generated by the leukocytes and VECs to elucidate the sequence of mechanical events involved in paracellular diapedesis. Our measurements suggest that the contractile stresses exerted by the leukocytes and the VECs can separately perturb the junctional tensions of VECs to result in the opening of gaps before the initiation of leukocyte transmigration. Decoupling the stresses exerted by the transmigrating leukocytes and the VECs reveals that the leukocytes actively contract the VECs to open a junctional gap and then push themselves across the gap by generating strong stresses that push into the matrix. In addition, we found that diapedesis is facilitated when the tension fluctuations in the VEC monolayer were increased by proinflammatory thrombin treatment. Our findings demonstrate that diapedesis can be mechanically regulated by the transmigrating leukocytes and by proinflammatory signals that increase VEC contractility.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Leukocytes/metabolism , Models, Biological , Transendothelial and Transepithelial Migration/physiology , HL-60 Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , Leukocytes/cytology
5.
Proc Natl Acad Sci U S A ; 115(50): E11681-E11690, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478057

ABSTRACT

The dramatic reorganization of chromatin during mitosis is perhaps one of the most fundamental of all cell processes. It remains unclear how epigenetic histone modifications, despite their crucial roles in regulating chromatin architectures, are dynamically coordinated with chromatin reorganization in controlling this process. We have developed and characterized biosensors with high sensitivity and specificity based on fluorescence resonance energy transfer (FRET). These biosensors were incorporated into nucleosomes to visualize histone H3 Lys-9 trimethylation (H3K9me3) and histone H3 Ser-10 phosphorylation (H3S10p) simultaneously in the same live cell. We observed an anticorrelated coupling in time between H3K9me3 and H3S10p in a single live cell during mitosis. A transient increase of H3S10p during mitosis is accompanied by a decrease of H3K9me3 that recovers before the restoration of H3S10p upon mitotic exit. We further showed that H3S10p is causatively critical for the decrease of H3K9me3 and the consequent reduction of heterochromatin structure, leading to the subsequent global chromatin reorganization and nuclear envelope dissolution as a cell enters mitosis. These results suggest a tight coupling of H3S10p and H3K9me3 dynamics in the regulation of heterochromatin dissolution before a global chromatin reorganization during mitosis.


Subject(s)
Biosensing Techniques/methods , Chromatin Assembly and Disassembly , Histone Code , Bacterial Proteins , Chromatin Assembly and Disassembly/genetics , Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins , HEK293 Cells , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Code/genetics , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Luminescent Proteins , Mitosis , Models, Biological , Single-Cell Analysis/methods
6.
Arterioscler Thromb Vasc Biol ; 39(12): 2492-2504, 2019 12.
Article in English | MEDLINE | ID: mdl-31597449

ABSTRACT

OBJECTIVE: Understanding message delivery among vascular cells is essential for deciphering the intercellular communications in cardiovascular diseases. MicroRNA (miR)-92a is enriched in endothelial cells (ECs) and circulation under atheroprone conditions. Macrophages are the primary immune cells in atherosclerotic lesions that modulate lesion development. Therefore, we hypothesize that, in response to atheroprone stimuli, ECs export miR-92a to macrophages to regulate their functions and enhance atherosclerotic progression. Approach and Results: We investigated the macrophage functions that are regulated by EC miR-92a under atheroprone microenvironments. We first determined the distributions of functional extracellular miR-92a by fractionating the intravesicular and extravesicular compartments from endothelial conditioned media and mice serum. The results indicate that extracellular vesicles are the primary vehicles for EC miR-92a transportation. Overexpression of miR-92a in ECs enhanced the proinflammatory responses and low-density lipoprotein uptake, while impaired the migration, of cocultured macrophage. Opposite effects were found in macrophages cocultured with ECs with miR-92a knockdown. Further analyses demonstrated that intravesicular miR-92a suppressed the expression of target gene KLF4 (Krüppel-like factor 4) in macrophages, suggesting a mechanism by which intravesicular miR-92a regulates recipient cell functions. Indeed, the overexpression of KLF4 rescued the EC miR-92a-induced macrophage atheroprone phenotypes. Furthermore, an inverse correlation of intravesicular miR-92a in blood serum and KLF4 expression in lesions was observed in atherosclerotic animals, indicating the potential function of extracellular miR-92a in regulating vascular diseases. CONCLUSIONS: EC miR-92a can be transported to macrophages through extracellular vesicles to regulate KLF4 levels, thus leading to the atheroprone phenotypes of macrophage and, hence, atherosclerotic lesion formation.


Subject(s)
Atherosclerosis/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Macrophages/metabolism , MicroRNAs/genetics , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Communication , Cells, Cultured , Extracellular Fluid/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/ultrastructure , Mice , MicroRNAs/biosynthesis , Microscopy, Electron, Transmission
7.
Proc Natl Acad Sci U S A ; 114(31): 8271-8276, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716920

ABSTRACT

Vascular endothelial cells (ECs) at arterial branches and curvatures experience disturbed blood flow and induce a quiescent-to-activated phenotypic transition of the adjacent smooth muscle cells (SMCs) and a subsequent smooth muscle hyperplasia. However, the mechanism underlying the flow pattern-specific initiation of EC-to-SMC signaling remains elusive. Our previous study demonstrated that endothelial microRNA-126-3p (miR-126-3p) acts as a key intercellular molecule to increase turnover of the recipient SMCs, and that its release is reduced by atheroprotective laminar shear (12 dynes/cm2) to ECs. Here we provide evidence that atherogenic oscillatory shear (0.5 ± 4 dynes/cm2), but not atheroprotective pulsatile shear (12 ± 4 dynes/cm2), increases the endothelial secretion of nonmembrane-bound miR-126-3p and other microRNAs (miRNAs) via the activation of SNAREs, vesicle-associated membrane protein 3 (VAMP3) and synaptosomal-associated protein 23 (SNAP23). Knockdown of VAMP3 and SNAP23 reduces endothelial secretion of miR-126-3p and miR-200a-3p, as well as the proliferation, migration, and suppression of contractile markers in SMCs caused by EC-coculture. Pharmacological intervention of mammalian target of rapamycin complex 1 in ECs blocks endothelial secretion and EC-to-SMC transfer of miR-126-3p through transcriptional inhibition of VAMP3 and SNAP23. Systemic inhibition of VAMP3 and SNAP23 by rapamycin or periadventitial application of the endocytosis inhibitor dynasore ameliorates the disturbed flow-induced neointimal formation, whereas intraluminal overexpression of SNAP23 aggravates it. Our findings demonstrate the flow-pattern-specificity of SNARE activation and its contribution to the miRNA-mediated EC-SMC communication.


Subject(s)
Hyperplasia/pathology , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , Animals , Endothelial Cells/physiology , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , Myocytes, Smooth Muscle/physiology , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 3/genetics
8.
Proc Natl Acad Sci U S A ; 113(41): 11525-11530, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27671657

ABSTRACT

The focal nature of atherosclerotic lesions suggests an important role of local hemodynamic environment. Recent studies have demonstrated significant roles of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in mediating mechanotransduction and vascular homeostasis. The objective of this study is to investigate the functional role of YAP/TAZ in the flow regulation of atheroprone endothelial phenotypes and the consequential development of atherosclerotic lesions. We found that exposure of cultured endothelial cells (ECs) to the atheroprone disturbed flow resulted in YAP/TAZ activation and translocation into EC nucleus to up-regulate the target genes, including cysteine-rich angiogenic inducer 61 (CYR61), connective tissue growth factor (CTGF), and ankyrin repeat domain 1 (ANKRD1). In contrast, the athero-protective laminar flow suppressed YAP/TAZ activities. En face analysis of mouse arteries demonstrated an increased nuclear localization of YAP/TAZ and elevated levels of the target genes in the endothelium in atheroprone areas compared with athero-protective areas. YAP/TAZ knockdown significantly attenuated the disturbed flow induction of EC proliferative and proinflammatory phenotypes, whereas overexpression of constitutively active YAP was sufficient to promote EC proliferation and inflammation. In addition, treatment with statin, an antiatherosclerotic drug, inhibited YAP/TAZ activities to diminish the disturbed flow-induced proliferation and inflammation. In vivo blockade of YAP/TAZ translation by morpholino oligos significantly reduced endothelial inflammation and the size of atherosclerotic lesions. Our results demonstrate a critical role of the activation of YAP/TAZ by disturbed flow in promoting atheroprone phenotypes and atherosclerotic lesion development. Therefore, inhibition of YAP/TAZ activation is a promising athero-protective therapeutic strategy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Rheology , Animals , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Carotid Artery, Common/pathology , Cell Cycle , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Connective Tissue Growth Factor/metabolism , Cysteine-Rich Protein 61/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/pathology , Mice , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Phenotype , Repressor Proteins/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
9.
Proc Natl Acad Sci U S A ; 113(8): 2206-11, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858399

ABSTRACT

The functional maturation and preservation of hepatic cells derived from human induced pluripotent stem cells (hiPSCs) are essential to personalized in vitro drug screening and disease study. Major liver functions are tightly linked to the 3D assembly of hepatocytes, with the supporting cell types from both endodermal and mesodermal origins in a hexagonal lobule unit. Although there are many reports on functional 2D cell differentiation, few studies have demonstrated the in vitro maturation of hiPSC-derived hepatic progenitor cells (hiPSC-HPCs) in a 3D environment that depicts the physiologically relevant cell combination and microarchitecture. The application of rapid, digital 3D bioprinting to tissue engineering has allowed 3D patterning of multiple cell types in a predefined biomimetic manner. Here we present a 3D hydrogel-based triculture model that embeds hiPSC-HPCs with human umbilical vein endothelial cells and adipose-derived stem cells in a microscale hexagonal architecture. In comparison with 2D monolayer culture and a 3D HPC-only model, our 3D triculture model shows both phenotypic and functional enhancements in the hiPSC-HPCs over weeks of in vitro culture. Specifically, we find improved morphological organization, higher liver-specific gene expression levels, increased metabolic product secretion, and enhanced cytochrome P450 induction. The application of bioprinting technology in tissue engineering enables the development of a 3D biomimetic liver model that recapitulates the native liver module architecture and could be used for various applications such as early drug screening and disease modeling.


Subject(s)
Bioprinting/methods , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Liver/anatomy & histology , Printing, Three-Dimensional , Albumins/biosynthesis , Biomimetics/methods , Cell Culture Techniques , Cell Differentiation , Gene Expression , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Liver/cytology , Tissue Engineering/methods
10.
Physiol Genomics ; 49(7): 339-345, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28500253

ABSTRACT

The long noncoding RNAs (lncRNAs), which constitute a large portion of the transcriptome, have gained intense research interest because of their roles in regulating physiological and pathophysiological functions in the cell. We identified from RNA-Seq profiling a set of lncRNAs in cultured human umbilical vein endothelial cells (HUVECs) that are differentially regulated by atheroprotective vs. atheroprone shear flows. Among the comprehensively annotated lncRNAs, including both known and novel transcripts, LINC00341 is one of the most abundant lncRNAs in endothelial cells. Moreover, its expression level is enhanced by atheroprotective pulsatile shear flow and atorvastatin. Overexpression of LINC00341 suppresses the expression of vascular cell adhesion molecule 1 (VCAM1) and the adhesion of monocytes induced by atheroprone flow and tumor necrosis factor-alpha. Underlying this anti-inflammatory role, LINC00341 guides enhancer of zest homolog 2, a core histone methyltransferase of polycomb repressive complex 2, to the promoter region of the VCAM1 gene to suppress VCAM1. Network analysis reveals that the key signaling pathways (e.g., Rho and PI3K/AKT) are co-regulated with LINC00341 in endothelial cells in response to pulsatile shear. Together, these findings suggest that LINC00341, as an example of lncRNAs, plays important roles in modulating endothelial function in health and disease.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Inflammation/genetics , RNA, Long Noncoding/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Atorvastatin/pharmacology , Cell Adhesion , Gene Expression Regulation , Gene Regulatory Networks , Humans , Inflammation/pathology , Monocytes/pathology , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Proc Natl Acad Sci U S A ; 111(28): 10347-52, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24958852

ABSTRACT

Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.


Subject(s)
Cardiovascular System/embryology , Embryonic Development/physiology , Endothelial Cells/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Animals , Cardiovascular System/cytology , Endothelial Cells/cytology , Ion Channels/genetics , Mice , Mice, Transgenic
12.
Proc Natl Acad Sci U S A ; 110(32): 13174-9, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23878263

ABSTRACT

Endothelial cells (ECs) are constantly exposed to xenobiotics and endobiotics or their metabolites, which perturb EC function, as well as to shear stress, which plays a crucial role in vascular homeostasis. Pregnane X receptor (PXR) is a nuclear receptor and a key regulator of the detoxification of xeno- and endobiotics. Here we show that laminar shear stress (LSS), the atheroprotective flow, activates PXR in ECs, whereas oscillatory shear stress, the atheroprone flow, suppresses PXR. LSS activation of PXR in cultured ECs led to the increased expression of a PXR target gene, multidrug resistance 1 (MDR1). An in vivo study using rats showed that the expression of MDR1 was significantly higher in the endothelium from the descending thoracic aorta, where flow is mostly laminar, than from the inner curvature of aortic arch, where flow is disturbed. Functionally, LSS-activated PXR protects ECs from apoptosis triggered by doxorubicin via the induction of MDR1 and other detoxification genes. PXR also suppressed the expression of proinflammatory adhesion molecules and monocyte adhesion in response to TNF-α and lipopolysaccharide. Overexpression of a constitutively active PXR in rat carotid arteries potently attenuated proinflammatory responses. In addition, cDNA microarray revealed a large number of the PXR-activated endothelial genes whose products are responsible for major steps of detoxification, including phase I and II metabolizing enzymes and transporters. These detoxification genes in ECs are induced by LSS in ECs in a PXR-dependent manner. In conclusion, our results indicate that PXR represents a flow-activated detoxification system to protect ECs against damage by xeno- and endobiotics.


Subject(s)
Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/metabolism , Inactivation, Metabolic/genetics , Receptors, Steroid/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Apoptosis/drug effects , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Blotting, Western , Carotid Arteries/metabolism , Cell Line, Tumor , Cells, Cultured , Cytochrome P-450 CYP1B1 , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Pregnane X Receptor , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stress, Mechanical , Xenobiotics/metabolism , Xenobiotics/pharmacology
13.
Stroke ; 46(4): 1099-106, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712942

ABSTRACT

BACKGROUND AND PURPOSE: Endovascular thrombectomy has shown promise for the treatment of acute strokes resulting from large-vessel occlusion. Reperfusion-related injury may contribute to the observed decoupling of angiographic and clinical outcomes. Iatrogenic disruption of the endothelium during thrombectomy is potentially a key mediator of this process that requires further study. METHODS: An in vitro live-cell platform was developed to study the effect of various commercially available endovascular devices on the endothelium. In vivo validation was performed using porcine subjects. RESULTS: This novel in vitro platform permitted high-resolution quantification and characterization of the pattern and timing of endothelial-cell injury among endovascular thrombectomy devices and vessel diameters. Thrombectomy devices displayed heterogeneous effects on the endothelium; the device performance assessed in vitro was substantiated by in vivo findings. CONCLUSIONS: In vitro live-cell artificial vessel modeling enables a detailed study of the endothelium after thrombectomy and may contribute to future device design. Large animal studies confirm the relevance of this in vitro system to investigate endothelial physiology. This artificial vessel model may represent a practical, scalable, and physiologically relevant system to assess new endovascular technologies.


Subject(s)
Endothelium, Vascular/injuries , Mechanical Thrombolysis , Stroke/therapy , Animals , Disease Models, Animal , In Vitro Techniques , Mechanical Thrombolysis/adverse effects , Mechanical Thrombolysis/instrumentation , Mechanical Thrombolysis/standards , Swine
14.
Circ Res ; 113(1): 40-51, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23603512

ABSTRACT

RATIONALE: Endothelial microRNA-126 (miR-126) modulates vascular development and angiogenesis. However, its role in the regulation of smooth muscle cell (SMC) function is unknown. OBJECTIVE: To elucidate the role of miR-126 secreted by endothelial cells (ECs) in regulating SMC turnover in vitro and in vivo, as well as the effects of shear stress on the regulation. METHODS AND RESULTS: Coculture of SMCs with ECs or treatment of SMCs with conditioned media from static EC monoculture (EC-CM) increased SMC miR-126 level and SMC turnover; these effects were abolished by inhibition of endothelial miR-126 and by the application of laminar shear stress to ECs. SMC miR-126 did not increase when treated with EC-CM from ECs subjected to inhibition of miR biogenesis, or with CM from sheared ECs. Depletion of extracellular/secreted vesicles in EC-CM did not affect the increase of SMC miR-126 by EC-CM. Biotinylated miR-126 or FLAG (DYKDDDDK epitope)-tagged Argonaute2 transfected into ECs was detected in the cocultured or EC-CM-treated SMCs, indicating a direct EC-to-SMC transmission of miR-126 and Argonaute2. Endothelial miR-126 represses forkhead box O3, B-cell lymphoma 2, and insulin receptor substrate 1 mRNAs in the cocultured SMCs, suggesting the functional roles of the transmitted miR-126. Systemic depletion of miR-126 in mice inhibited neointimal lesion formation of carotid arteries induced by cessation of blood flow. Administration of EC-CM or miR-126 mitigated the inhibitory effect. CONCLUSIONS: Endothelial miR-126 acts as a key intercellular mediator to increase SMC turnover, and its release is reduced by atheroprotective laminar shear stress.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/physiology , Myocytes, Smooth Muscle/cytology , Animals , Argonaute Proteins/genetics , Argonaute Proteins/physiology , Carotid Artery, Common/pathology , Cell Culture Techniques/instrumentation , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Silencing , Genes, bcl-2 , Hemorheology , Human Umbilical Vein Endothelial Cells , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Ligation , Mice , MicroRNAs/genetics , MicroRNAs/therapeutic use , Muscle, Smooth, Vascular/cytology , Neointima , Paracrine Communication , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Recombinant Fusion Proteins/physiology , Umbilical Arteries/cytology
15.
Arterioscler Thromb Vasc Biol ; 34(10): 2191-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24876354

ABSTRACT

Atherosclerosis develops preferentially at branches and curvatures of the arterial tree, where blood flow pattern is disturbed rather than being laminar, and wall shear stress has an irregular distribution without defined directions. The endothelium in the atherosusceptible regions, in comparison to that in atheroresistant regions, shows activation of proproliferative and proinflammatory gene expressions, reduced production of nitric oxide (NO), increased leukocyte adhesion, and permeability, as well as other atheroprone phenotypes. Differences in gene expressions and cell phenotypes have been detected in endothelia residing in native atherosusceptible and atheroresistant regions of the arteries, or in arteries from animal models with artificial creation of disturbed flow. Similar results have also been shown in in vitro systems that apply controlled shear stresses with or without clear directions to cultured endothelial cells in fluid-dynamically designed flow-loading devices. The available evidence indicates that the coordination of multiple signaling networks, rather than individual separate pathways, links the mechanical signals to specific genetic circuitries in orchestrating the mechanoresponsive networks to evoke comprehensive genetic and functional responses.


Subject(s)
Atherosclerosis/pathology , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Mechanotransduction, Cellular , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Hemodynamics , Humans , Phenotype , Regional Blood Flow , Stress, Mechanical
16.
Arterioscler Thromb Vasc Biol ; 34(7): 1437-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24855060

ABSTRACT

OBJECTIVE: The site-specificity of endothelial phenotype is attributable to the local hemodynamic forces. The flow regulation of microRNAs in endothelial cells (ECs) plays a significant role in vascular homeostasis and diseases. The objective of this study was to elucidate the molecular mechanism by which the pulsatile shear flow-induced microRNA-23b (miR-23b) exerts antiproliferative effects on ECs. APPROACH AND RESULTS: We used a combination of a cell perfusion system and experimental animals to examine the flow regulation of miR-23b in modulating EC proliferation. Our results demonstrated that pulsatile shear flow induces the transcription factor Krüppel-like factor 2 to promote miR-23b biosynthesis; the increase in miR-23b then represses cyclin H to impair the activity and integrity of cyclin-dependent kinase-activating kinase (CAK) complex. The inhibitory effect of miR-23b on CAK exerts dual actions to suppress cell cycle progression, and reduce basal transcription by deactivating RNA polymerase II. Whereas pulsatile shear flow regulates the miR-23b/CAK pathway to exert antiproliferative effects on ECs, oscillatory shear flow has little effect on the miR-23b/CAK pathway and hence does not cause EC growth arrest. Such flow pattern-dependent phenomena are validated with an in vivo model on rat carotid artery: the flow disturbance induced by partial carotid ligation led to a lower expression of miR-23b and a higher EC proliferation in comparison with the pulsatile flow regions of the unligated vessels. Local delivery of miR-23b mitigated the proliferative EC phenotype in partially ligated vessels. CONCLUSIONS: Our findings unveil a novel mechanism by which hemodynamic forces modulate EC proliferative phenotype through the miR-23b/CAK pathway.


Subject(s)
Carotid Artery Diseases/enzymology , Cell Proliferation , Cyclin H/metabolism , Cyclin-Dependent Kinases/metabolism , Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , MicroRNAs/metabolism , Transcription, Genetic , Animals , Carotid Artery Diseases/genetics , Carotid Artery Diseases/physiopathology , Cell Cycle Checkpoints , Cells, Cultured , Cyclin H/genetics , Cyclin-Dependent Kinases/genetics , Disease Models, Animal , Humans , Kruppel-Like Transcription Factors/metabolism , Male , Mechanotransduction, Cellular , MicroRNAs/genetics , Perfusion , Phenotype , Pulsatile Flow , RNA Interference , RNA Polymerase II/metabolism , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Stress, Mechanical , Time Factors , Transfection , Cyclin-Dependent Kinase-Activating Kinase
17.
Proc Natl Acad Sci U S A ; 109(28): 11110-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22665785

ABSTRACT

We use a novel 3D inter-/intracellular force microscopy technique based on 3D traction force microscopy to measure the cell-cell junctional and intracellular tensions in subconfluent and confluent vascular endothelial cell (EC) monolayers under static and shear flow conditions. We found that z-direction cell-cell junctional tensions are higher in confluent EC monolayers than those in subconfluent ECs, which cannot be revealed in the previous 2D methods. Under static conditions, subconfluent cells are under spatially non-uniform tensions, whereas cells in confluent monolayers are under uniform tensions. The shear modulations of EC cytoskeletal remodeling, extracellular matrix (ECM) adhesions, and cell-cell junctions lead to significant changes in intracellular tensions. When a confluent monolayer is subjected to flow shear stresses with a high forward component comparable to that seen in the straight part of the arterial system, the intracellular and junction tensions preferentially increase along the flow direction over time, which may be related to the relocation of adherens junction proteins. The increases in intracellular tensions are shown to be a result of chemo-mechanical responses of the ECs under flow shear rather than a direct result of mechanical loading. In contrast, the intracellular tensions do not show a preferential orientation under oscillatory flow with a very low mean shear. These differences in the directionality and magnitude of intracellular tensions may modulate translation and transcription of ECs under different flow patterns, thus affecting their susceptibility for atherogenesis.


Subject(s)
Endothelial Cells/cytology , Animals , Atherosclerosis , Cell Communication , Cells, Cultured/cytology , Extracellular Matrix/metabolism , Finite Element Analysis , Fluorescence Resonance Energy Transfer , Humans , Imaging, Three-Dimensional , Microscopy, Atomic Force/methods , Microscopy, Confocal/methods , Models, Biological , Models, Statistical , Oscillometry/methods , Shear Strength
18.
Proc Natl Acad Sci U S A ; 107(22): 10268-73, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20479254

ABSTRACT

Shear stress imposed by blood flow is crucial for maintaining vascular homeostasis. We examined the role of shear stress in regulating SIRT1, an NAD(+)-dependent deacetylase, and its functional relevance in vitro and in vivo. The application of laminar flow increased SIRT1 level and activity, mitochondrial biogenesis, and expression of SIRT1-regulated genes in cultured endothelial cells (ECs). When the effects of different flow patterns were compared in vitro, SIRT1 level was significantly higher in ECs exposed to physiologically relevant pulsatile flow than pathophysiologically relevant oscillatory flow. These results are in concert with the finding that SIRT1 level was higher in the mouse thoracic aorta exposed to atheroprotective flow than in the aortic arch under atheroprone flow. Because laminar shear stress activates AMP-activated protein kinase (AMPK), with subsequent phosphorylation of endothelial nitric oxide synthase (eNOS) at Ser-633 and Ser-1177, we studied the interplay of AMPK and SIRT1 on eNOS. Laminar flow increased SIRT1-eNOS association and eNOS deacetylation. By using the AMPK inhibitor and eNOS Ser-633 and -1177 mutants, we demonstrated that AMPK phosphorylation of eNOS is needed to prime SIRT1-induced deacetylation of eNOS to enhance NO production. To verify this finding in vivo, we compared the acetylation status of eNOS in thoracic aortas from AMPKalpha2(-/-) mice and their AMPKalpha2(+/+) littermates. Our finding that AMPKalpha2(-/-) mice had a higher eNOS acetylation indicates that AMPK phosphorylation of eNOS is required for the SIRT1 deacetylation of eNOS. These results suggest that atheroprotective flow, via AMPK and SIRT1, increases NO bioavailability in endothelium.


Subject(s)
Blood Vessels/physiology , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Acetylation , Amino Acid Substitution , Animals , Aorta/physiology , Base Sequence , Cells, Cultured , DNA Primers/genetics , Endothelial Cells/physiology , Enzyme Activation , Hemorheology , Homeostasis , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mutagenesis, Site-Directed , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pulsatile Flow , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stress, Mechanical
19.
Proc Natl Acad Sci U S A ; 107(7): 3240-4, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133739

ABSTRACT

Endothelial cells (ECs) respond to changes in mechanical forces, leading to the modulation of signaling networks and cell function; an example is the inhibition of EC proliferation by steady laminar flow. MicroRNAs (miRs) are short noncoding 20-22 nucleotide RNAs that negatively regulate the expression of target genes at the posttranscriptional level. This study demonstrates that miRs are involved in the flow regulation of gene expression in ECs. With the use of microRNA chip array, we found that laminar shear stress (12 dyn/cm(2), 12 h) regulated the EC expression of many miRs, including miR-19a. We further showed that stable transfection of miR-19a significantly decreased the expression of a reporter gene controlled by a conserved 3'-untranslated region of the cyclinD1 gene and also the protein level of cyclin D1, leading to an arrest of cell cycle at G1/S transition. Laminar flow suppressed cyclin D1 protein level, and this suppressive effect was diminished when the endogenous miR-19a was inhibited. In conclusion, we demonstrated that miR-19a plays an important role in the flow regulation of cyclin D1 expression. These results revealed a mechanism by which mechanical forces modulate endothelial gene expression.


Subject(s)
Cyclin D1/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Regional Blood Flow/physiology , Umbilical Veins/cytology , Base Pairing , Base Sequence , Biomechanical Phenomena , Blotting, Western , DNA Primers/genetics , Flow Cytometry , Humans , MicroRNAs/genetics , Microarray Analysis , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Umbilical Veins/physiology
20.
Proc Natl Acad Sci U S A ; 107(7): 3234-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133741

ABSTRACT

MicroRNAs (miRs) can regulate many cellular functions, but their roles in regulating responses of vascular endothelial cells (ECs) to mechanical stimuli remain unexplored. We hypothesize that the physiological responses of ECs are regulated by not only mRNA and protein signaling networks, but also expression of the corresponding miRs. EC growth arrest induced by pulsatile shear (PS) flow is an important feature for flow regulation of ECs. miR profiling showed that 21 miRs are differentially expressed (8 up- and 13 downregulated) in response to 24-h PS as compared to static condition (ST). The mRNA expression profile indicates EC growth arrest under 24-h PS. Analysis of differentially expressed miRs yielded 68 predicted mRNA targets that overlapped with results of microarray mRNA profiling. Functional analysis of miR profile indicates that the cell cycle network is highly regulated. The upregulation of miR-23b and miR-27b was found to correlate with the PS-induced EC growth arrest. Inhibition of miR-23b using antagomir-23b oligonucleotide (AM23b) reversed the PS-induced E2F1 reduction and retinoblastoma (Rb) hypophosphorylation and attenuated the PS-induced G1/G0 arrest. Antagomir AM27b regulated E2F1 expression, but did not affect Rb and growth arrest. Our findings indicate that PS suppresses EC proliferation through the regulation of miR-23b and provide insights into the role of miRs in mechanotransduction.


Subject(s)
Endothelial Cells/physiology , Gene Expression Regulation/physiology , MicroRNAs/physiology , Pulsatile Flow/physiology , Retinoblastoma Protein/metabolism , Analysis of Variance , Bromodeoxyuridine , Cell Proliferation , Flow Cytometry , Humans , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides/genetics , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL